TECHNICAL FIELD
[0001] The invention relates to the field of biomedicine. In particular, the invention relates
to molecules that are antagonists of human microRNAs miR-100, miR-20, miR-222, miR-181,
and miR-92, and to the use of same as a drug, preferably as a drug for the treatment
of myotonic dystrophy type 1.
BACKGROUND OF THE INVENTION
[0002] Muscular dystrophy (MD) represents a group of over 30 genetic diseases characterized
by a progressive weakness and degeneration of the skeletal muscles that control movement.
Some forms of MD present in infancy or childhood, while others may not appear until
middle age or later. The prognosis for people with MD varies depending on the type
and progression of the disorder. Some cases may be mild and progress very slowly over
a normal lifetime, while others result in severe muscle weakness, functional disability,
and loss of the ability to walk. Within muscular dystrophies, a distinction is made
between those in which patients have problems relaxing the muscles after a voluntary
contraction, known as myotonia. There are two types of myotonic dystrophy, type 1
(DM1) and type 2 (DM2).
[0003] DM1 is further classified into three subtypes that sometimes have signs and symptoms
in common: mild, classic, and congenital (present at birth). Symptoms of the mild
form are the least severe, with a normal life expectancy. The classic form is characterized
by muscle weakness and atrophy, inability of the muscles to relax quickly after contracting
(myotonia), cataracts, and abnormal heart function. Some adults with the classic form
may have a physical disability. Finally, the congenital form is characterized by severe
generalized weakness at birth (hypotonia), which results in problems with breathing
and can lead to premature death.
[0004] DM1 is caused by mutations in the
DMPK gene and is inherited in an autosomal dominant manner. In particular, these mutations
correspond to the expansion of a CTG trinucleotide repeat in the 3' non-coding region
of the
DMPK gene. A number of repeats greater than 34 CTG is considered abnormal, and molecular
genetic testing detects pathogenic variants in almost 100% of affected individuals.
Expression of expanded alleles in DM1 results in nuclear retention of mutant DMPK
mRNA. The mutant transcripts sequester splicing factors that are similar to the
Drosophila Muscleblind-like protein (MBNL), resulting in abnormal alternative splicing of a
multitude of other transcripts and expression of fetal forms of the corresponding
proteins in adults with DM1. The
Drosophila Muscleblind-like protein has vertebrate homologs called MBNL1-3, which are also known
for their ability to regulate splicing. MBNL1 is strongly expressed in skeletal and
cardiac muscle tissue and during myoblast differentiation. Its expression is lower
in other tissues such as brain, placenta, lungs, liver, kidney, and pancreas. MBNL2
has a largely overlapping expression and is detected in heart, brain, placenta, lungs,
liver, skeletal muscle, kidneys, and pancreas. In turn, MBNL3 is expressed in placenta
and satellite cells.
[0005] Although abnormalities in the splicing mechanism are considered the main factor underlying
the pathogenesis of DM1, it has been shown that other mechanisms may also be involved
in this disease, such as additional changes in gene expression, antisense transcripts,
sequestration of other RNA-binding proteins or miRNAs, translation efficiency, alternative
polyadenylation dysregulation, and miRNA dysregulation.
[0006] Currently there is no specific treatment for progressive weakness in people with
DM1 and, therefore, it is of interest to explore new therapeutic strategies. Among
the therapeutic approaches carried out in animal models of DM1, the most interesting
results derive from blocking the interaction between MBNLs and toxic RNA using small
molecules, peptides, morpholinos or antisense oligonucleotides, and gapmers to degrade
mutant
DMPK transcripts. However, a less explored alternative in DM1 is the therapeutic modulation
of
MBNL1 and
MBNL2 gene expression, in particular the overexpression of said genes to increase the levels
of said proteins, especially in tissues where they are normally transcribed. It is
further preferred that said increased levels occur in at least one or more of the
relevant tissues and organs where particularly significant symptoms of the disease
appear, such as skeletal and smooth muscle, heart, and nervous system. In particular,
the authors of the present invention have previously described that antagonists against
human microRNAs miR-23-3p and miR-218-5p have therapeutic potential in the treatment
of DM1 by being able to modulate endogenous MBNL proteins (patent
ES2659845B1).
[0007] Therefore, given that microRNAs (commonly abbreviated as miRs) are known to be involved
in numerous important biological processes such as metabolism, cell cycle, migration,
epithelial-mesenchymal transition (EMT), cell differentiation, and survival, and pathological
processes such as cancer, and since their regulation can be achieved with the use
of antagonist molecules, the present invention describes antagonists of microRNAs
miR-100, miR-20, miR-222, miR-181, and miR-92 to increase MBNL1 and MBNL2 protein
levels, and proposes the use of such inhibitors in the treatment of DM1
SUMMARY OF THE INVENTION
[0008] In one aspect, the present invention relates to an antagonist of human microRNA miR-100
capable of increasing the intracellular levels of MBNL1 and/or MBNL2 proteins, for
use thereof in the treatment of myotonic dystrophy type 1. Preferably, the intracellular
levels of MBNL1 and/or MBNL2 proteins are increased in a muscle cell, preferably in
a muscle cell having a muscular dystrophy phenotype, preferably muscular dystrophy
type 1. In a preferred embodiment, the antagonist is a molecule of an oligonucleotide
nature and/or an oligonucleotide analog, preferably an antimiR, a blockmiR, or a microRNA
sponge.
[0009] Preferably, the antagonist comprises a region of at least 7 contiguous nucleotide
or nucleotide analog units which is at least 100% identical to the complementary sequence
of SEQ ID NO: 1.
[0010] Preferably, the antagonist comprises a region of at least 15 contiguous nucleotide
or nucleotide analog units which is at least 90% identical to the sequence complementary
to any region present in SEQ ID NO: 1.
[0011] Preferably, the antagonist comprises a region of at least 7 contiguous nucleotide
or nucleotide analog units which is 100% identical to the sequence complementary to
the seed region of human microRNA miR-100 as defined in SEQ ID NO: 2.
[0012] Preferably, the antagonist comprises a first region of at least 7 contiguous nucleotide
or nucleotide analog units which is 100% identical to the sequence complementary to
the seed region of human microRNA miR-100 as defined in SEQ ID NO: 2, and wherein
said antagonist further comprises a second region of at least 7 contiguous nucleotide
or nucleotide analog units adjacent to the first region, wherein said second region
is at least 90% identical to the sequence complementary to any region present in SEQ
ID NO: 1.
[0013] Preferably, the antagonist comprises a region of at least 7 contiguous nucleotide
or nucleotide analog units which is at least 95% identical to SEQ ID NO: 3. Preferably,
the antagonist comprises a region of at least 15 contiguous nucleotide or nucleotide
analog units which is 100% identical to any region present in SEQ ID NO: 3. Preferably,
the antagonist consists of SEQ ID NO: 3.
[0014] Preferably, the antagonist:
- i) comprises a region which is at least 95% identical to SEQ ID NO: 3,
- ii) comprises at least one nucleotide in turn comprising one or more chemical modifications
in the ribose moiety, in the phosphate bond, or in both, and
- iii) optionally has at the 5' end and/or at the 3' end one or more additional molecules,
wherein preferably said one or more additional molecules are not deoxyribonucleotide
or ribonucleotide derivatives, and wherein preferably it is cholesterol.
[0015] Preferably, the antagonist comprises a region of at least 8 contiguous nucleotide
or nucleotide analog units which is at least 95% identical to any region present in
SEQ ID NO: 8, 13, or 26. Preferably, the antagonist comprises a region of at least
15 contiguous nucleotide or nucleotide analog units which is at least 95% identical
to any region present in SEQ ID NO: 8, 13, or 26. Preferably, the antagonist consists
of SEQ ID NO: 8, 13, or 26.
[0016] In another aspect, the present invention relates to a composition comprising at least
one antagonist of human microRNA miR-100 as defined in the preceding aspect or any
of the preferred embodiments thereof, for use thereof in the treatment of myotonic
dystrophy type 1. Preferably, said composition is a pharmaceutical composition further
comprising a pharmaceutically acceptable vehicle and/or one or more pharmaceutically
acceptable excipients.
BRIEF DESCRIPTION OF THE FIGURES
[0017]
Figure 1: Expression levels of genes of interest in HeLa cells. Base-2 logarithmic representation (log2) of the relative expression level of the
of MBNL1 (left) and MBNL2 (right) transcripts in HeLa cells treated with (A) the mimetics from the initial
screening performed with the SureFind Transcriptome PCR Array miRNA plates and (B)
selection of those microRNAs of interest, as well as miR-23b and -218 as a comparison.
For both genes, the reference value is that detected in HeLa cells treated without
any mimetic and the endogenous gene used to normalize is GAPDH. *p<0.05 ,**p<0.01,***p<0.001
(Student's t-test).
Figure 2: Toxicity measurements in the cell model of the disease. Graphical representation of the percentage of cell viability inhibition at 72 h,
obtained in patient cells caused by toxicity associated with a dose-response assay
with increasing amounts of the antagomiRs miR100, miR-20a, miR-181c, miR-222, and
miR-92a, with respect to the base-10 logarithm of the nanomolar concentration of the
compound. The amounts tested in the cells are indicated at the bottom of the corresponding
curves. The dotted line indicates the dose corresponding to TC50. Z-factor of the
assay: 0.64. Symbols represent the mean ± SEM.
Figure 3: Levels of protein of interest in the cell model of the disease. Relative quantification of the levels of MBNL1 protein, measured by means of Quantitative
Dot Blot, in DM1 muscle cells transfected with the concentrations and antagomiRs indicated
and compared with the levels detected in control muscle cells (CNT). Bars represent
the mean ± SEM. GAPDH was used as the endogenous control to normalize expression.
* p<0.05, ** p<0.01, *** p<0.001 (one-way ANOVA test)
Figure 4. Evaluation of the activity and toxicity of the different antagomiRs in the cell
model of the disease. Representation of the toxicity (percentage of cell viability inhibition, black line)
and the activity (percentage increase in the expression of MBNL1, gray line and bars)
in DM1 cells after transfection with the different antagomiRs at the different concentrations
tested (each in triplicate). Each symbol and bar represents the mean ± SEM
Figure 5: Expression of the microRNAs of interest in different tissues. Expression levels of miR-23b-3p, miR-218-5p-1, miR-218-5p-2, miR-20a-5p, miR-92a-3p,
miR-100-5p, miR-181c-5p, and miR-222-3p in transcripts per million (TPMs), in the
different tissues of interest: trachea, thyroid, testicles, stomach, spleen, different
types of muscle cells, skeletal muscle, liver, kidney, heart, bladder, esophagus,
diaphragm, colon, cervix, brain, and blood. The data is from the sequencing of 100
samples.
Figure 6: Demonstration of the overexpression of the different miRNAs in a cell model of the
disease. The expression levels of the miRNAs indicated in DM1 muscle cells and controls from
Dr. Denis Furling (Arandel et al. 2017) are shown. These levels have been quantified by quantitative PCR and have been
normalized to the expression of endogenous controls U1, U6, and miR-103. Bars represent
the mean ± SEM. p<0.05 ,**p<0.01,***p<0.001 (Student's t-test).
DEFINITIONS
[0018] It should be noted that, as used herein, the singular forms "a", "an", and "the"
include plural references unless the context clearly indicates otherwise. Furthermore,
unless otherwise indicated, the term "at least" preceding a series of elements should
be understood to refer to each element in the series. Those skilled in the art will
recognize, or will be able to determine using no more than routine experimentation,
many equivalents to the specific embodiments of the invention described herein. It
is intended that said equivalents be encompassed by the present invention.
[0019] As used herein, the combined term "and/or" among multiple listed elements is understood
to encompass both individual and combined options. For example, when two elements
are joined by "and/or", a first option refers to the applicability of the first element
without the second element. A second option refers to the applicability of the second
element without the first element. A third option refers to the applicability of the
first and second elements together. It is understood that any of these options fall
within the meaning and, therefore, satisfy the requirement of the term "and/or" as
used herein. It is also understood that the simultaneous applicability of more than
one of the options falls within the meaning and, therefore, satisfies the requirement
of the term "and/or".
[0020] Throughout this specification and the claims that follow, unless the context requires
otherwise, the word "comprises" will be understood to imply the inclusion of an integer
or step or group of integers or steps, but not the exclusion of any other integer
or step or group of integers or steps. As used herein, the term "comprising" may be
replaced with the term "containing" or "including" or "having". The terms "comprising",
"containing", "including", "having", whenever used herein in the context of an aspect
or embodiment of the present invention, may be replaced with the term "consisting
of", although this is less preferred. As used herein, "consisting of" excludes any
element, step, or ingredient not specified in the claim element.
[0021] The term "hybridization" is used to refer to the structure formed by 2 or more independent
RNA or DNA strands forming a double or triple stranded structure by means of pairing
the bases of one strand to the other. These base pairs are considered G-C, A-U/T,
and G-U/T. (A-Adenine, C-Cytosine, G-Guanine, U-Uracil, T-Thymine). As in the case
of complementarity, hybridization can be full or partial.
[0022] The terms "complementary" and "complementarity" are interchangeable and refer to
the ability of oligonucleotides to form base pairs with each other. Base pairs are
normally formed by means of hydrogen bonds between nucleotide units in antiparallel
polynucleotide strands or regions. Complementary polynucleotide strands or regions
can form base pairs, for example, as in the way of Watson-Crick (e.g., A to T, A to
U, C to G). A 100% (or perfect) complementarity refers to the situation in which each
nucleotide unit of one oligonucleotide strand or region can form hydrogen bonds or
bridges with each nucleotide unit of a second polynucleotide region or strand. Partial
or non-perfect complementarity refers to the situation in which some, but not all,
nucleotide units of two strands or two regions can form hydrogen bonds with each other
and can be expressed as a percentage. The terms "sequence identity" or "percent identity"
in the context of two or more nucleotides, polypeptides, or proteins refer to two
or more sequences or subsequences that are the same ("identical") or have a specific
percentage of nucleobases or amino acids that are identical ("percent identity") when
compared and aligned to obtain maximum correspondence with a second molecule, as measured
using a local sequence comparison algorithm (e.g., by means of BLAST alignment or
any other local algorithm known to skilled persons), or alternatively, by visual inspection.
It should be noted that percent identity as used here is measured in the context of
a local alignment, i.e., it is based on the alignment of regions of local similarity
between sequences, unlike a global alignment, the objective of which is to align two
sequences over their entire length. Therefore, in the context of the present invention,
percent identity is calculated solely on the basis of a local alignment comparison
algorithm.
[0023] In the context of the present invention, complementarity is considered to occur under
stringent hybridization conditions. "Stringent hybridization conditions" refers to
conditions in which nucleotides that have homology with the target sequence preferably
hybridize with the target sequence, and nucleotides that do not have homology with
the target sequence do not substantially hybridize. The stringent conditions depend
on the sequence, as longer sequences hybridize specifically at higher temperatures.
Generally speaking, stringent conditions are chosen to be approximately 5°C lower
than the thermal melting temperature (Tm) of the specific sequence. Tm is the temperature
at which 50% of the nucleotides complementary to the target sequence hybridize with
the target sequence in equilibrium at a prescribed ionic strength, pH, and nucleic
acid concentration.
[0024] "Fragment" or "region" of a sequence is understood to mean a portion of a polypeptide
or nucleic acid molecule preferably containing at least 10%, 20%, 30%, 40%, 50%, 50%,
60%, 70%, 80%, 90%, 95%, or more of the total length of the reference nucleic acid
molecule or polypeptide. A fragment may contain at least 5, 6, 7, 8, 8, 9, 10, 10,
11, 11, 12, 13, 13, 14, 14, 15, 16, 17, 18, 19, or 20 nucleotides with respect to
the reference sequence.
[0025] "MBNL1" is understood to mean the Muscleblind-like Splicing Regulator 1 protein encoded
by the
MBNL1 gene. The term "MBNL1" as used in the present description includes all variants encoded
by this gene.
[0026] "MBNL2" is understood to mean the Muscleblind-like Splicing Regulator 2 protein encoded
by the
MBNL2 gene. The term "MBNL2" as used in the present description includes all variants encoded
by this gene.
[0027] "Myotonic dystrophy" (DM) refers to a group of inherited disorders called muscular
dystrophies. There are two main types of myotonic dystrophy: type 1 (DM1) and type
2 (DM2). Myotonic dystrophy type 1 (DM1), as defined herein, is a multisystem disorder
that affects skeletal and smooth muscle, as well as other tissues, such as the eye,
heart, endocrine system, and central nervous system. The clinical manifestations of
DM1 span a continuum from mild to severe and have been broadly categorized into three
phenotypes: mild, classic, and congenital. Mild DM1 is characterized by cataracts
and mild myotonia (sustained muscle contraction); life expectancy is normal. Classic
DM1 is characterized by muscle weakness and atrophy, myotonia, cataracts, and often
cardiac conduction abnormalities; adults may become physically disabled and may have
a shorter life span. Congenital DM1 is characterized by hypotonia and severe generalized
weakness at birth, often with respiratory failure and premature death; intellectual
disability is common.
[0028] "Treatment of myotonic dystrophy" refers to the treatment of symptomatic or asymptomatic
myotonic dystrophy, at least partially restoring the phenotype of a patient with such
a disease to a non-pathological state. "Prevention of myotonic dystrophy" involves
measures taken to prevent the development of myotonic dystrophy (i.e., preventing
the onset of histopathology and/or symptomatology-associated myotonic dystrophy).
[0029] An "effective dose" or "therapeutically effective dose" is an amount sufficient to
achieve a beneficial or desired clinical outcome. In the context of the present invention,
the following definitions of TC50, EC50, Emax, and Tlndex have been applied:
TC50 refers to the dose at which antagomiR produces toxic effects (lack of cell viability)
in 50 percent of the cells on which it is being tested in a 72-hour period.
[0030] EC50 refers to the concentration of the antagomiR required to reach 50 percent of
its maximum activity in a 72-hour period.
[0031] Emax refers to the maximum activity (in this case, expression of MBNL1 or MBNL2 proteins
measured in fold change) of the antagomiR in a 72-hour period.
[0032] Tindex refers to a measure of a drug's safety margin and efficacy. It is expressed
numerically as a ratio between the concentration of the antagomiR that causes cell
death (TC50) taking into account its Emax and the dose that causes the desired therapeutic
effect (EC50).
[0033] "Expression vector" means a nucleic acid molecule (usually a plasmid), a virus, an
RNA molecule, a minichromosome, liposome, exosome, or a cell, designed to express
or deliver a specific biomolecule, in the present case an antagonist. The expression
vector is introduced into a host cell using well-known techniques such as infection
or transfection, including calcium phosphate transfection, liposome-mediated transfection,
electroporation, and sonoporation. The expression constructs and methods for their
generation and use to express a desired protein are known to one skilled in the art.
DETAILED DESCRIPTION OF THE INVENTION
[0034] It is widely accepted that myotonic dystrophy type 1 (DM1) is caused, among other
molecular defects, by the sequestration and consequent lack of function of MBNL1 and
2 proteins, which are abnormally sequestered by CUG expansions in disease-causing
mutant
DMPK transcripts. In the present invention, the inventors set out to identify the miRNAs
that endogenously repress the expression of MBNL1 and 2 in order to, by means of inhibitors
thereof (antagomiRs and blockmiRs), find those molecules that could prevent such inhibition
and thereby increase the levels of MBNL1 and 2 proteins, the function of which is
reduced under pathological conditions. AntagomiR and antimiR are used synonymously
in the present description.
[0035] For this purpose, a screening was performed in HeLa cells in which a set of 90 agomiRs
(miRNA mimetics) were overexpressed and the levels of
MBNL1 and
MBNL2 transcripts were quantified by means of real-time PCR. A total of 5 miRNAs were found
to be capable of repressing the expression of MBNL1 and/or MBNL2 under these conditions:
hsa-miR-20a-5p (SEQ ID NO:4), hsa-miR-181c-5p (SEQ ID NO: 5), hsa-miR-222-3p (SEQ
ID NO:6), hsa-miR-100-5p (SEQ ID NO:3), hsa-miR-92a-3p (SEQ ID NO: 7). Next, antisense
oligonucleotides were designed against each of the identified miRNAs, known as antagomiRs,
and tested in patient-derived muscle cells. The relative quantification of MBNL1 protein
levels can be seen in Figure 3, wherein it is shown that while most cause only less
notable increases, some antagomiRs are active at lower concentrations than others,
for example, antagomiR-20a or - 100, which at 10 nM already recovers to normal levels
(see control), and others are able to double expression in DM1 cells (antagomiR-92a).
[0036] Then, the inventors proceeded to evaluate toxicity in the cell model of the disease.
Table 3 shows the TC50, EC50, Emax, and Tlndex values of the antagonists described
in the present invention, wherein a very high Tlndex can be observed in the case of
antagomiR-100, derived from its low toxicity (TC50) and EC50.
[0037] In view of these above results, and taking into account that other antagonists that
raise MBNL1 levels and are used for the treatment of DM1 (
ES2659845B1) are known in the prior art, it can be concluded that antagonists of miR-100-5p,
miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p have significant therapeutic potential
by increasing MBNL1 levels and can be used as drugs, particularly in the treatment
of patients with DM1. Furthermore, results were also obtained on the ability of these
antagonists to decrease the relative expression levels of MBNL2, as shown in Figure
1.
[0038] Therefore, a
first aspect of the invention relates to an antagonist of a human microRNA, wherein said human
microRNA is selected from the group of miR-100, miR-20a, miR-222, miR-181c, and miR-92a,
and wherein said antagonist is capable of increasing the intracellular levels of the
MBNL1 and/or MBNL2 proteins, and/or the intracellular levels of the transcripts (mRNA)
of the genes encoding the MBNL1 and/or MBNL2 proteins. In a preferred embodiment,
the antagonist of a human microRNA is an antagonist of human microRNA miR-100.
[0039] In another preferred embodiment, the antagonist of human microRNA miR-100 is capable
of increasing the intracellular levels of the MBNL1 and/or MBNL2 proteins and/or the
intracellular levels of the transcripts of the genes encoding the MBNL1 and/or MBNL2
proteins in a muscle cell, preferably in a muscle cell having a muscular dystrophy
phenotype, preferably type 1. As used herein, the expression "capable of increasing
the levels of the MBNL1 and/or MBNL2 proteins or their transcripts" refers to an increase,
preferably a statistically significant increase, in the intracellular levels of said
proteins (or any of their variants) or in the intracellular levels of the mRNA encoding
them, caused by treatment with the antagonists of the present invention, wherein said
increase is in comparison to the levels of a control or untreated cell or cell population.
Control cell or cell population refers to cells not treated with the antagonists of
the invention and having an abnormally reduced MBNL1 and/or MBNL2 function due to
muscular dystrophy type 1 or due to the presence of at least one of the miRNAs acting
negatively on their expression. In statistics, a result or effect is statistically
significant when it is unlikely to have been due to chance. The significance level
of a statistical test is a statistical concept associated with the verification of
a hypothesis. Said test is defined as the probability of making the decision to reject
the null hypothesis when it is true (a decision known as type I error, or false positive).
The decision is often made using the p-value: if the p-value is below the significance
level, then the null hypothesis is rejected. The lower the p-value, the more significant
the result. Preferably, increased intracellular levels of proteins or their transcripts
are considered to be statistically significant compared to the levels of said proteins
or transcripts in control cell(s) when the p-significance levels are equal to or less
than 0.05, 0.01, and 0.001.
[0040] Preferably, the control cells are muscle cells having a muscular dystrophy type 1
phenotype. Preferably, the intracellular levels of MBNL1 and/or MBNL2 proteins and/or
transcripts thereof increase by at least 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2,
3, 4, 5, 6, 7, 8, 9, 10, or more fold changes with respect to the levels of said proteins/transcripts
present in a control or untreated cell or cells .
[0041] In that sense, the present inventors propose the modulation of endogenous MBNL proteins
by causing the sequestration of at least one of the miRNAs that act negatively on
their expression, thereby giving rise to an up-regulation and, as a consequence, to
an increase in the intracellular levels of endogenous MBNL1 and/or MBNL2 proteins
or their transcripts (mRNAs). Thus, the aim is to modulate the endogenous protein
by means of silencing or decreasing the activity of human microRNAs miR-100, miR-20a,
miR-222, miR-181c, or miR-92a.
[0042] As used herein, "antagonist" refers to any molecule capable of blocking the action
of human microRNAs, preferably miR-100, miR-20a, miR-222, miR-18c1, and miR-92a, i.e.,
compounds, regardless of their nature, that are capable of producing a decrease in
the endogenous activity of said miRNAs, and thereby increasing the intracellular levels
of MBNL1 and/or MBNL2 proteins and/or their transcripts. Since the present invention
focuses on decreasing the activity of human microRNAs miR-100, miR-20a, miR-222, miR-181c,
and miR-92a, said repressor capacity will be diminished by the presence of the inhibitors,
silencers, or blockers thereof, which are all referred to as antagonists. Therefore,
similarly, the effect produced by an inhibitor, silencer, or blocker entails and signifies
an antagonism of the action of the same. Preferably, the miR-100, miR-20a, miR-222,
miR-181c, and miR-92a the action of which is blocked by the antagonist of the present
invention are specifically miR-100-5p, miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p.
[0043] In one embodiment, the antagonist capable of blocking the action of human microRNA,
preferably miR-100, miR-20a, miR-222, miR-18c1, and miR-92a, is a molecule, preferably
a low molecular weight organic compound.
[0044] In a preferred embodiment, the antagonist is a molecule of an oligonucleotide nature
and/or an oligonucleotide analog. As used in the present specification, the terms
"molecule of an oligonucleotide nature" or "oligonucleotide" refers to molecules resulting
from the binding of usually not more than 50 units of the monomers that give rise
to the molecules known in abbreviated form as DNA or RNA, which are monomers or oligomers
made up of a phosphate group, the nitrogenous bases adenine (A), cytosine (C), guanine
(G), or uracil (U) or thymine (T), and the pentose known as ribose (in the case of
oligoribonucleotides in the case of RNA) or deoxyribose (in the case of oligodeoxyribonucleotides,
or DNA). Oligodeoxynucleotides (DNA) and oligoribonucleotides (RNA) are therefore
considered to be included among oligonucleotides. By common usage, molecules the units
of which include the nucleotide inosine and molecules that are a mixture of DNA/RNA
because they contain regions of both types are also considered to be included within
said definition.
[0045] In a preferred embodiment, the antagonist is a molecule of an oligoribonucleotide
nature (a oligoribonucleotide) and/or an oligoribonucleotide analog. As used in the
present specification, the terms "molecule of an oligoribonucleotide nature" or "oligoribonucleotide"
refer to molecules resulting from the binding of usually not more than 50 units of
the monomers that give rise to the molecule known in abbreviated form as RNA, which
are monomers made up of a phosphate group, the nitrogenous bases adenine (A), cytosine
(C), guanine (G), or uracil (U), and pentose. By common usage, molecules the units
of which include the nucleotide inosine are also considered to be included within
said definition.
[0046] As used in the present invention, the terms "oligonucleotide analog" and "oligoribonucleotide
analog" encompass molecules derived from oligonucleotides and oligoribonucleotide,
respectively, incorporating artificially introduced chemical modifications, such as
those described in
Antisense part III: chemistries Aug 28, 2018 ericminikel • Cambridge, MA. (https://www.cureffi.org/2018/08/28/antisense-part-iii-chemistries/). Preferably, the modifications are among one of the following modifications: i)
a chemical modification in at least one of the component units, either in the phosphate
group, the pentose, or one of the nitrogenous bases; or ii) replacement of one or
more of the basic components constituting the oligonucleotide with a different chemical
structure but with the same function. In the case of ii), these include, without limitation,
the use of morpholino rings replacing pentoses to arrange the nitrogenous bases in
their suitable spatial arrangement, or the replacement of phosphodiester internucleotide
bonds with phosphorothioate internucleotide bonds. Furthermore, other modifications
iii) also include those consisting of the addition of groups of a non-nucleotide nature
at the 5' and/or 3' ends; and/or iv) the addition of further oligonucleotide sequences
or oligonucleotide analogs (i.e., not complementary to the target microRNA) at said
ends. Some of the common chemical modifications by which oligonucleotide and oligoribonucleotide
molecules are modified include modification of part or all of the nucleotides with
2'-methoxy group (2'-O-methyl: 2'-OMe), 2'-O-methoxyethyl group (2'-MOE), and/or phosphorothioate
bonds.
[0047] In particular, for the purposes of the invention, of special interest (and considered
to be included within the modifications that give rise to antagonists included within
the scope of the invention) are those modifications, valid for oligonucleotides and
oligoribonucleotides that give rise to DNA and RNA analogs with increased resistance
to hydrolysis, and which are generally modifications on the pentose, such as those
resulting in: 2'-O-methyl-substituted (the 2'-methoxy modifications); 2'-O-methoxyethyl-substituted;
LNAs (locked nucleic acids, in which the pentose moiety is modified with an extra
bridge connecting the 2' oxygen and 4' carbon and locking the pentose in the 3'-endo
conformation); BNAs (bridged nucleic acids); PMOs (nucleic acids wherein the ribose
has been replaced by a morpholino group); or PNAs (peptide nucleic acids, wherein
the pentose phosphate group is replaced by an amino acid residue, such that the nucleotide
analog has as its backbone a structure of repeating N-(2-aminoethyl)-glycine units
attached by peptide bonds). Recently, nucleotides with different modifications have
also been used, such as CRNs (conformationally restricted nucleotides), in which the
pentose moiety is locked in a conformation controlled by means of a chemical moiety
that acts as a connector, a modification that is being used mainly to obtain antagomiRs
with new properties, or cETs (cET = constrained ethyl).
[0048] Also included among the possible modifications that give rise to oligonucleotide
or oligoribonucleotide analogs of the invention are modifications that give rise to
phosphorothioate bonds, which are modifications that affect phosphate groups that
are part of the "backbone" of the nucleotide chain, giving rise to the introduction
of a sulfur atom replacing an oxygen atom of the phosphate group that is not acting
as a bridge between nucleotides; these modifications make the bonds between nucleotides
resistant to degradation by nucleases, so it is common to introduce them between the
last 3-5 nucleotides located at the 5' or 3' ends of the antagonist oligonucleotide
to inhibit degradation thereof by exonucleases, increasing stability. Phosphorothioate
bonds also appear to increase binding to serum proteins, such as albumins, to improve
the distribution of molecules containing them. Also included among the chemical modifications
giving rise to oligonucleotide and oligoribonucleotide analogs of the invention is
methylation at 5' of the nitrogenous base cytosine (C), which appears to increase
the stability of the complexes formed with the target.
[0049] Other chemical modifications, which are also comprised within the possible modifications
which give rise to oligonucleotide or oligoribonucleotide analogs, are also possible
and known. As can be deduced from the definition of "molecules of an oligonucleotide/oligoribonucleotide
nature" and of "oligonucleotide/oligoribonucleotide analogs", also comprised within
the definition of oligonucleotide or oligoribonucleotide analogs are molecules in
which some units have modifications and others do not, as well as hybrids between
nucleic acid analogs and peptides or even hybrid molecules in which some of the nucleotide
units are ribonucleotides (or analogs thereof) and others are deoxyribonucleotides
(nucleotides in which the sugar is deoxyribose), as well as analogs of the latter,
i.e., RNA-DNA hybrids and analogs thereof.
[0050] Preferably, the molecule of an oligonucleotide nature and/or an oligonucleotide analog
is an antagonist of a human microRNA selected from the group of miR-100, miR-20, miR-222,
miR-181, and miR-92, and wherein said antagonist is an antimiR, a blockmiR, or an
miRNA sponge.
[0051] As used herein, "blockmiR" refers to small oligonucleotides with a special chemistry
designed against the sequence that a particular miRNA, in this case miR-100-5p, miR-20a-5p,
miR-222-3p, miR-181c-5p, and miR-92a-3p, recognizes in its target messenger RNA (mRNA),
so each of them should only de-repress the effect of that miRNA on that transcript,
with a very specific effect being expected. Therefore, they are designed to have a
sequence that is complementary to that of a fragment of the sequence of an mRNA that
serves as a binding site for an miRNA, such that they typically bind in the 3' untranslated
region (3'-UTR) of an mRNA, i.e., in the area in which endogenous miRNAs typically
bind.
[0052] In a preferred embodiment, the antagonist is an antimiR. AntimiRs are generally used
to silence endogenous miRNAs. Therefore, as used herein, "antimiR", which is synonymous
with "antagomiR", refers to small synthetic oligonucleotides, preferably synthetic
oligoribonucleotides, that are complementary to a target miRNA, in this case miR-100-5p,
miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p, and therefore act as antagonists
of said miRNAs. Typically, antimiRs include chemical modifications such as 2-O-methyl
groups, phosphorothioates, and conjugated fatty acids, preferably cholesterol.
[0053] As used herein, "miRNA sponge" refers to molecules the main constituent of which
is oligonucleotide repeats arranged in tandem, with the particularity that each one
of said oligonucleotides is itself or contains a binding site for binding to the miR-100-5p,
miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p that it antagonizes.
[0054] To design antagonist molecules, it is important to take into account the existence
of sufficient complementarity with the sequence of the target miR, in the case of
antimiRs and miRNA sponges, or their target mRNA, in the case of blockmiRs, to which
they must bind in order to actually produce the desired inhibition/antagonism effect.
In that sense, examples of the "typical" complementarity between an antagonist and
its target being 50% can be taken into account. Therefore, in a preferred embodiment,
the antagonist of an oligonucleotide or oligoribonucleotide nature of the invention
comprises a fragment of the sequence of nucleotide or ribonucleotide units, or analog
units thereof, in which the sequence of the nitrogenous bases of the ribonucleotide
or ribonucleotide analog units is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%,
90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to the
complementary sequence of the endogenous molecule with which it is to hybridize, i.e.,
to the sequences of miR-100-5p, miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p
to which it must bind (in the case of antagomiRs and the repeat sequence of miRNA
sponges) or the sequence of the fragment of the messenger mRNA (in the case of blockmiRs).
In the context of the present invention, nitrogenous bases T and U are considered
interchangeable, and therefore the sequences including T can also be considered sequences
with U, and vice versa.
[0055] In the event that the antagonist of an oligonucleotide or oligoribonucleotide nature
of the invention is an antimiR, in a preferred embodiment, said antimiR comprises
a fragment of the sequence of nucleotide or ribonucleotide units, or analog units
thereof, in which the sequence of the nitrogenous bases of the ribonucleotide or ribonucleotide
analog units is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%,
94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to the complementary sequence
of any of sequences of human microRNAs SEQ ID NO: 1 (miR-100-5p), SEQ ID NO: 14 (miR-20a-5p),
SEQ ID NO: 15 (miR-222-3p), SEQ ID NO: 16 (miR-181c-5p), or SEQ ID NO: 17 (miR-92a-3p).
[0056] Furthermore, endogenous miRNAs usually bind in the 3' untranslated region (3'-UTR)
of the mRNA they regulate. However, the binding between the miRNA and its target mRNA
does not have to be perfect and is mainly dominated by the so-called seed region of
the miRNA. The seed region sequence is a conserved sequence that is mainly located
at positions 2-8 of the 5' end of the miRNA. Even if the base pairing of the miRNA
and its target mRNA does not perfectly match, with the "seed sequence" it has to perfectly
match, i.e., 100%, complementary. In the case of miR-100-5p, its sequence corresponds
to SEQ ID NO: 1, of which the seed region is made up of SEQ ID NO: 2. In the case
of miR-20a-5p, its sequence corresponds to SEQ ID NO: 14, of which the seed region
is made up of SEQ ID NO: 18. In the case of miR-222-3p, its sequence corresponds to
SEQ ID NO: 15, of which the seed region is made up of SEQ ID NO: 19. In the case of
miR-181c-5p, its sequence corresponds to SEQ ID NO: 16, of which the seed region is
made up of SEQ ID NO: 20. In the case of miR-92a-3p, its sequence corresponds to SEQ
ID NO: 17, of which the seed region is made up of SEQ ID NO: 21.
[0057] It can be deduced from the foregoing that when designing antagonists of human microRNAs
miR-100-5p, miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p of an oligonucleotide
or oligoribonucleotide nature or analogs thereof, it is advisable to consider the
seed region thereof, and to include the region highly complementary to said seed region
in the antagonist being designed. Therefore, in a preferred embodiment, the antagonist
of an oligonucleotide or oligoribonucleotide nature of the invention is an antimiR
comprising a fragment of the sequence of nucleotide or ribonucleotide units or analog
units thereof, in which the sequence of the nitrogenous bases of the ribonucleotide
or ribonucleotide analog units is at least 95%, 96%, 97%, 98%, 98%, 99%, or 99.5%,
or 100% identical to the sequence complementary to the seed region of any of human
microRNAs miR-100-5p, miR-20a-5p, miR-222-3p, miR-181c-5p, or miR-92a-3p, as determined
by SEQ ID NOs: 2, 18, 19, 20, 20, or 21, respectively. Preferably, the identity to
the sequence complementary to the seed region is 100%.
[0058] The length of the sequence of the antagonists of an oligonucleotide or oligoribonucleotide
nature or analogs thereof is at least 8 nucleotides, without a maximum having been
established. Preferably, the length of the sequence of said antagonists is at least
8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40,
45, or 50 nucleotides. In a preferred embodiment, the length of said antagonists is
from 10-15, 10-20, 20-30, 30-40, or 40-50. In another preferred embodiment, the length
of said antagonists is from 15-25 nucleotides. In one embodiment of the present invention,
the antagonist is an antimiR comprising or consisting of a region of at least 7, 8,
9, 10, 11, 12, 13, 14, 15, 20, 21, 22, or 23, preferably between 20 and 23, contiguous
nucleotide or nucleotide analog units which is at least 50%, 55%, 60%, 65%, 70%, 75%,
80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical
to the complementary sequence of any of sequences SEQ ID NO: 1 (miR-100-5p), SEQ ID
NO: 14 (miR-20a-5p), SEQ ID NO: 15 (miR-222-3p), SEQ ID NO: 16 (miR-181c-5p), or SEQ
ID NO: 17 (miR-92a-3p), preferably SEQ ID NO: 1. In a preferred embodiment, the antagonist
is an antimiR comprising or consisting of a region of at least 5, 6, 7, or 8 contiguous
nucleotide or nucleotide analog units which is 100% identical to the sequence complementary
to the seed region of any of human microRNAs as defined in SEQ ID NO: 2 (miR-100-5p
seed), SEQ ID NO: 18 (miR-20a-5p seed), SEQ ID NO: 19 (miR-222-3p seed), SEQ ID NO:
20 (miR-181c-5p seed), or SEQ ID NO: 21 (miR-92a-3p seed), preferably SEQ ID NO: 2.
[0059] In another preferred embodiment, the antagonist is an antimiR comprising a first
region of at least 5, 6, 7, or 8 contiguous nucleotide or nucleotide analog units
which is 100% identical to the sequence complementary to the seed region of any of
human microRNAs miR-100, miR-20, miR-222, miR-181, or miR-92, as defined in SEQ ID
NO: 2 (miR-100-5p seed), SEQ ID NO: 18 (miR-20a-5p seed), SEQ ID NO: 19 (miR-222-3p
seed), SEQ ID NO: 20 (miR-181c-5p seed), and SEQ ID NO: 21 (miR-92a-3p seed), and
wherein said antagonist further comprises a second region of at least 10, 11, 12,
13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 contiguous nucleotide or nucleotide
analog units adjacent to the first region, wherein said second region is at least
50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%,
99%, or 99.5%, or 100% identical to the sequence complementary to any region present
in SEQ ID NO: 1 (miR-100-5p), SEQ ID NO: 14 (miR-20a-5p), SEQ ID NO: 15 (miR-222-3p),
SEQ ID NO: 16 (miR-181c-5p), or SEQ ID NO: 17 (miR-92a-3p), respectively.
[0060] As described in the examples, the authors of the present invention found that antagonists
of human microRNAs miR-100-5p, miR-20a-5p, miR-222-3p, miR-181c-5p, and miR-92a-3p,
are capable of increasing the levels of the MBNL1 protein and the expression levels
of the MBNL2 protein in a muscle cell. Therefore, a series of antimiRs specific against
said miRs were designed. Human microRNA sequences, from 5' to 3', are (the seed region
is underlined):
hsa-miR-100-5p (SEQ ID NO: 1): AACCCGUAGAUCCGAACUUGUG
hsa-miR-20a-5p (SEQ ID NO: 14): UAAAGUGCUUAUAGUGCAGGUAG
hsa-miR-181c-5p (SEQ ID NO: 16): AACAUUCAACCUGUCGGUGAGU
hsa-miR-222-3p (SEQ ID NO: 15): AGCUACAUCUGGCUACUGGGU
hsa-miR-92a-3p (SEQ ID NO: 17): UAUUGCACUUGUCCCGGCCUGU
[0061] The sequences of the designed antagomiRs are, from 5' to 3':
AntagomiR-100-5p (SEQ ID NO: 3): CACAAGTTCGGATCTACGGGTT
AntagomiR-20a-5p (SEQ ID NO: 4): CTACCTGCACTATAAGCACTTTA
AntagomiR-181c-5p (SEQ ID NO: 5): ACTCACCGACAGGTTGAATGTT
AntagomiR-222-3p (SEQ ID NO: 6): ACCCAGTAGCCAGATGTAGCT
AntagomiR-92a-3p (SEQ ID NO: 7): ACAGGCCGGGACAAGTGCAATA
[0062] In the case of antimiRs, the antagonist of an oligonucleotide or oligoribonucleotide
nature of the invention comprises or consists of a fragment of the sequence of nucleotide
or ribonucleotide units, or analog units thereof, in which the sequence of the nitrogenous
bases of the nucleotide or nucleotide analog units is at least 50%, 55%, 60%, 65%,
70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or
100% identical to any of sequences SEQ ID NO: 3, 4, 5, 6, or 7, preferably SEQ ID
NO: 3. In a preferred embodiment, the antagonist consists of sequence SEQ ID NO: 3,
4, 5, 6, or 7, preferably SEQ ID NO: 3.
[0063] In one embodiment of the present invention, the antagonist is an antimiR comprising
or consisting of a region of at least 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 22,
or 23, preferably between 20 and 23, contiguous nucleotide or nucleotide analog units
which is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%,
95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any region present in any
of sequences SEQ ID NO: 3, 4, 5, 6, or 7, preferably SEQ ID NO: 3.
[0064] Furthermore, the developed antagomiRs can have certain chemical modifications that
are common in this type of oligonucleotide or oligoribonucleotide analogs, among which
are the following:
- 2'-O-methyl (2'-methoxy) modifications on all the pentose moieties,
- the replacement of some phosphate bonds between monomeric nucleotide analog units
with phosphorothioate bonds, and
- the incorporation of fatty acid molecules at one end of the molecule, preferably at
the 3' end.
[0065] In this way, in a preferred embodiment, the antagonist has at least one chemical
modification in its structure, wherein:
- i. said antagonist comprises a region of at least 7, 8, 9, 10, 11, 12, 13, 14, 15,
20, 21, 22, or 23, preferably between 20 and 23, contiguous nucleotide or nucleotide
analog units which is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any region present
in any of sequences SEQ ID NO: 3, 4, 5, 6, or 7, preferably SEQ ID NO: 3,
- ii. at least one of the nucleotide or nucleotide analog units making up said antagonist
is a nucleotide analog having one or more chemical modifications in the pentose moiety,
preferably ribose, in the phosphate bond, or in both, and
- iii. optionally, said antagonist has at the 5' end and/or at the 3' end one or more
additional molecules, preferably one or more molecules which are not deoxyribonucleotide
or ribonucleotide derivatives.
[0066] In another preferred embodiment, the antagonist has at least one chemical modification
in its structure, wherein:
- i. said antagonist comprises a region of at least 7, 8, 9, 10, 11, 12, 13, 14, 15,
20, 21, 22, or 23, preferably between 20 and 23, contiguous nucleotide or nucleotide
analog units which is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any region present
in any of sequences SEQ ID NO: 3, 4, 5, 6, or 7, preferably SEQ ID NO: 3,
- ii. at least one of the nucleotide or nucleotide analog units making up said antagonist
is a nucleotide analog having one or more chemical modifications selected from the
group of: methylations, phosphorothioate bonds, LNAs, BNAs, constrained ethyl (cEt)
nucleosides, PMOs, PNAs, 2'-O-methyl-substituted, 2'-O-methoxyethyl-substituted, 2'-fluoro,
and
- iii. optionally, said antagonist has at the 5' end and/or at the 3' end one or more
additional molecules, preferably one or more molecules which are not deoxyribonucleotide
or ribonucleotide derivatives.
[0067] In another preferred embodiment, the antagonist has at least one chemical modification
in its structure, wherein:
- i. said antagonist comprises a region of at least 7, 8, 9, 10, 11, 12, 13, 14, 15,
20, 21, 22, or 23, preferably between 20 and 23, contiguous nucleotide or nucleotide
analog units which is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any region present
in any of sequences SEQ ID NO: 3, 4, 5, 6, or 7, preferably SEQ ID NO: 3,
- ii. at least one of the nucleotide or nucleotide analog units making up said antagonist
is a nucleotide analog having one or more chemical modifications in the ribose moiety,
in the phosphate bond, or in both, preferably selected from the group of: methylations,
phosphorothioate bonds, LNAs, BNAs, constrained ethyl (cEt) nucleosides, PMOs, PNAs,
2'-O-methyl-substituted, 2'-O-methoxyethyl-substituted, 2'-fluoro, and
- iii. optionally, said antagonist has at the 5' end and/or at the 3' end one or more
additional molecules, preferably one or more molecules which are not deoxyribonucleotide
or ribonucleotide derivatives, preferably cholesterol.
[0068] This therefore gives rise to the following oligonucleotide or oligoribonucleotide
analogs antagonists of human microRNAs miR-100, miR-20, miR-222, miR-181, and miR-92:
AntagomiR-100-5p (SEQ ID NO: 8): C*A*CAAGTTCGGATCTACGG*G*T*T*
AntagomiR-20a-5p (SEQ ID NO: 9): C*T*ACCTGCACTATAAGCACT*T*T*A*
AntagomiR-181c-5p (SEQ ID NO: 10): A*C*TCACCGACAGGTTGAAT*G*T*T*
AntagomiR-222-3p (SEQ ID NO: 11): A*C*CCAGTAGCCAGATGTA*G*C*T*
AntagomiR-92a-3p (SEQ ID NO: 12): A*C*AGGCCGGGACAAGTGCA*A*T*A*
where:
- an asterisk (*) indicates the presence of a phosphorothioate bond,
- A, C, T, G in bold indicates that said nucleotide is modified with 2'-O-methoxyethyl,
and
- optionally, a fatty acid is conjugated at the 3' and/or 5' ends of the molecule.
[0069] Therefore, all the nitrogenous bases of sequences SEQ ID NO: 8, 9, 10, 11, and 12
are modified with 2'-O-methoxyethyl (2'-OMe).
[0070] In a preferred aspect, sequences SEQ ID NO: 8, 9, 10, 11, and 12 have a fatty acid,
preferably cholesterol, conjugated at their 3' end.
AntagomiR-100-5p (SEQ ID NO: 13): C*A*CAAGTTCGGATCTACGG*G*T*T*-Chol
AntagomiR-20a-5p (SEQ ID NO: 22): C*T*ACCTGCACTATAAGCACT*T*T*A*-Chol
AntagomiR-181c-5p (SEQ ID NO: 23): A*C*TCACCGACAGGTTGAAT*G*T*T*-Chol
AntagomiR-222-3p (SEQ ID NO: 24): A*C*CCAGTAGCCAGATGTA*G*C*T*-Chol
AntagomiR-92a-3p (SEQ ID NO: 25): A*C*AGGCCGGGACAAGTGCA*A*T*A*-Chol
where:
- an asterisk (*) indicates the presence of a phosphorothioate bond,
- A, C, T, G in bold indicates that said nucleotide is modified with 2'-O-methoxyethyl,
and
- "-Chol" indicates that the oligonucleotide is conjugated to cholesterol at its 3'
end.
[0071] Therefore, all the nitrogenous bases of sequences SEQ ID NO: 13, 22, 23, 24, and
25 are modified with 2'-O-methoxyethyl (2'-OMe) and conjugated a cholesterol.
[0072] In a preferred embodiment, the antagonist of an oligonucleotide or oligoribonucleotide
nature of the invention is an antimiR comprising or consisting of a fragment of the
sequence of nucleotide or ribonucleotide units, or analog units thereof, in which
the sequence of the nitrogenous bases of the nucleotide or nucleotide analog units
is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%,
96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any of the sequences SEQ ID NO:
8, 9, 10, 11, or 12, preferably SEQ ID NO: 8. In a preferred embodiment, the antagonist
consists of SEQ ID NO: 8, 9, 10, 11, or 12, preferably SEQ ID NO: 8.
[0073] In one embodiment of the present invention, the antagonist is an antimiR comprising
or consisting of a region of at least 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 22,
or 23, preferably between 20 and 23, contiguous nucleotide or nucleotide analog units
which is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%,
95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any of the sequences SEQ ID
NO: 8, 9, 10, 11, or 12, preferably SEQ ID NO: 8.
[0074] In a preferred embodiment, the antagonist of an oligonucleotide or oligoribonucleotide
nature of the invention is an antimiR comprising or consisting of a fragment of the
sequence of nucleotide or ribonucleotide units, or analog units thereof, in which
the sequence of the nitrogenous bases of the nucleotide or nucleotide analog units
is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%,
96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any of the sequences SEQ ID NO:
13, 22, 23, 24, 25 or 26-30, preferably SEQ ID NO: 13; wherein a cholesterol molecule
is conjugated at the 3' end of the antagonist. In a preferred embodiment, the antagonist
consists of any of the sequences SEQ ID NO: 13, 22, 23, 24, 25 or 26-30, preferably
SEQ ID NO: 13, wherein a cholesterol molecule is conjugated at the 3' end of said
antagonist.
[0075] In one embodiment of the present invention, the antagonist is an antimiR comprising
or consisting of a region of at least 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 21, 22,
or 23, preferably between 20 and 23, contiguous nucleotide or nucleotide analog units
which is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%,
95%, 96%, 97%, 98%, 99%, or 99.5%, or 100% identical to any of the sequences SEQ ID
NO: 13, 22, 23, 24, 25 or 26-30, preferably SEQ ID NO: 13, wherein a cholesterol molecule
is conjugated at the 3' end of said antagonist.
[0076] A possible embodiment of the invention can be considered to be a molecule of an oligonucleotide
nature and/or an oligonucleotide analog which is an antagonist of a human microRNA,
wherein said human microRNA is selected from the group of miR-100, miR-20, miR-222,
miR-181, and miR-92, or a mixture of two or more of said molecules, which is capable
of increasing the intracellular levels of MBNL1 and/or MBNL2 proteins or the levels
of their transcripts in a muscle cell. Said muscle cell can be one or more cells of
the skeletal muscle tissue, cardiac muscle tissue, or smooth muscle tissue, or the
precursors thereof. In a preferred embodiment, said muscle cell is a cardiomyocyte,
myoblast, myocyte, striated muscle cell, or smooth muscle cell. In another preferred
embodiment, the antagonist is capable of increasing the levels of MBNL1 and/or MBNL2
proteins or the intracellular levels of their transcripts in a cell found in one or
more organs selected from the group of brain, cerebellum, hippocampus, or other organ
of the central nervous system, skeletal muscle, heart, adipose tissue, kidney, liver
and biliary system, lung, pharynx, nasopharynx, nose, placenta, spleen, testicle,
uterus, gastrointestinal tract, breast, bladder, prostate, skin, or in one or more
cells from a primary culture of one of said organs or from an established cell line
derived from one of said organs (including induced pluripotent stem cells, known as
iPSCs) or stem cells from one of said organs.
[0077] In a second aspect, the present invention relates to a composition comprising at
least one antagonist as defined in the first aspect or in any of its embodiments,
and at least one pharmaceutically acceptable excipient. Furthermore, the antagonist
can be delivered in a vector. Said vector can be an expression vector. The vector
can be selected from the list including DNA, RNA, viral, liposome, or exosome vectors.
The present invention thereby also includes any delivery or expression means comprising
the inhibitors or antagonists defined in the first aspect or in any of its embodiments.
[0078] It is common to prepare compositions comprising more than one active ingredient.
For example, it would be possible to prepare a composition comprising the combination
of the antagonists of the present invention with other antagonists of other human
microRNAs which also intervene or have an effect on the production of MBNL proteins.
In that sense, in a preferred embodiment of the second aspect, the composition comprises
a combination of antagonists, which include the antagonist as defined in the first
aspect or in any of its embodiments. In a preferred aspect, the composition according
to the second aspect comprises at least one antagonist as defined in the first aspect
or in any of its embodiments, and at least one antagonist of human microRNA-218-5p
or human microRNA-23b-3p. Human microRNA-218-5p or human microRNA-23b-3p are defined
in patent
ES 2 659 845 A1. In another embodiment, the composition according to the second aspect comprises
at least one antagonist as defined in the first aspect or in any of its embodiments,
and other molecules of a nucleotide nature, preferably antisense oligonucleotides
(ASOs).
[0079] Preferably, the composition according to the second aspect comprises at least one
antagonist against human microRNA miR-100, preferably an antagonist comprising or
consisting of SEQ ID NO: 8, 13, or 26, or an antagonist the sequence of which is at
least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,
98%, 99%, or 99.5%, or 100% identical to any of the sequences SEQ ID NO: 8, 13, or
26.
[0080] For clinical application, the compositions of the present invention, which will then
be considered pharmaceutical compositions of the present invention, can be prepared
in a form suitable for the desired application. As disclosed in publications also
relating to the clinical application of inhibitors/antagonists of microRNAs, such
as international application
WO2012148373A1, this will generally involve the preparation of compositions that are essentially
free of pyrogens, as well as other impurities that may be harmful to humans or animals.
Said international application
WO2012148373A1 relates to compounds analogous to those of the present invention and to therapeutic
applications thereof, whereby information on forms of preparation and presentation
of pharmaceutical compositions, possible suitable administration vehicles, or forms
and routes of administration may be considered applicable to the present invention
and may be taken as a reference for the compositions of the present invention.
[0081] Colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres,
beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles,
and liposomes, can be used as delivery vehicles for the inhibitors/antagonists of
the present invention, with which the pharmaceutical composition of the invention
is formed, but the possibility for the antagonists to be administered without the
aid of other pharmacological agents is also included.
[0082] Another possibility, as already discussed, is to prepare the pharmaceutical compositions
of the invention using suitable salts and buffers to make the delivery vehicles stable
and aid in capture by the target cells. The compositions of the present invention
may be aqueous compositions comprising an effective amount of the delivery vehicle
and comprising either the molecules of an oligonucleotide nature of the invention,
independently or forming liposomes or other complexes, or expression vectors thereof,
dissolved or dispersed in a pharmaceutically acceptable vehicle or aqueous medium.
The expressions "pharmaceutically acceptable" or "pharmacologically acceptable" refer
to molecular entities and compositions that do not produce adverse reactions, allergic
or otherwise, when administered to an animal or human being. As used in the present
specification, "pharmaceutically acceptable vehicle" includes solvents, buffers, solutions,
dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption
retarding agents, and the like, acceptable for use in formulating pharmaceuticals,
such as pharmaceuticals suitable for administration to humans. The use of such media
and agents for pharmaceutically active substances is well known in the art. Except
to the extent that any conventional medium or agent is incompatible with the active
ingredients of the present invention, the use thereof in the pharmaceutical compositions
of the invention is contemplated. Supplemental active ingredients may also be incorporated
in the compositions, provided that they do not inactivate the molecules of the present
invention or their expression vectors.
[0083] The active compositions of the present invention can be administered by any of the
common routes, provided that the target tissue is available through that route. This
includes oral, nasal, or buccal routes and also, preferably, administration may be
by intradermal, subcutaneous, intramuscular, intraperitoneal, or intravenous routes.
[0084] Suitable pharmaceutical forms for injectable use include, for example, sterile aqueous
solutions or sterile dispersions and powders for the extemporaneous preparation of
sterile injectable solutions or dispersions. Generally, these preparations are sterile
and fluid to the extent that there is easy injectability. The preparations must be
stable under manufacturing and storage conditions and must be preserved against the
contaminating action of microorganisms, such as bacteria and fungi. Suitable solvents
or dispersion media may contain, for example, water, ethanol, polyol (e.g., glycerol,
propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures
thereof, and vegetable oils. Suitable fluidity can be maintained, for example, by
means of using a coating, such as lecithin, by means of maintaining the required particle
size in the case of dispersion, and by means of using surfactants. Prevention of the
action of microorganisms can be brought about by various antibacterial and antifungal
agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and
the like. In many cases, it will be preferable to include isotonic agents, for example,
sugars or sodium chloride. Prolonged absorption of the injectable compositions can
be caused by use in the compositions of agents that delay absorption, for example,
aluminum monostearate and gelatin.
[0085] Sterile injectable solutions can be prepared by incorporating the active compounds
in a suitable amount in a solvent along with any other ingredients (e.g., as specified
above) as desired, followed by sterilization by filtration. Generally, dispersions
are prepared by incorporating the various sterilized active ingredients in a sterile
vehicle containing the basic dispersion medium and the other desired ingredients,
e.g., as specified above. In the case of sterile powders for the preparation of sterile
injectable solutions, preferred methods of preparation include vacuum drying and lyophilization,
techniques that produce a powder of the active ingredient(s) plus any additional desired
ingredients from a solution thereof previously sterilized by filtration.
[0086] The compositions of the present invention can generally be formulated in a neutral
or salt form. Pharmaceutically acceptable salts include, for example, acid addition
salts (formed with the free amino groups of the protein) derived from inorganic acids
(e.g., hydrochloric or phosphoric acid), or from organic acids (e.g., acetic, oxalic,
tartaric, mandelic acids), and the like. Salts formed with the free carboxyl groups
of the protein can also be derived from inorganic bases (e.g., sodium, potassium,
ammonium, calcium, or ferric hydroxides) or organic bases (e.g., isopropylamine, trimethylamine,
histidine, procaine, and the like).
[0087] In any case, it is recommended that the preparation of the compositions of the present
invention follow practices that ensure a minimum quality for use in humans, such as
those contained in the Guide to Good Manufacturing Practice for Active Pharmaceutical
Ingredients of Working Group Q7 of the International Conference on Harmonization of
Technical Requirements for Registration of Pharmaceuticals for Human Use.
[0088] Preferably, other quality guidelines from the same source, such as Q8 on Pharmaceutical
Development or Q10 on the Pharmaceutical Quality System, are also taken into account.
[0089] After formulation, the solutions are preferably administered in a form compatible
with the dosage formulation and in such an amount as to be therapeutically effective.
The formulations can be readily administered in a variety of dosage forms such as
injectable solutions, drug release capsules, and the like. For parenteral administration
in an aqueous solution, for example, the solution is generally suitably buffered and
the liquid diluent must first be made isotonic, for example, with sufficient saline
or glucose. Such aqueous solutions can be used, for example, for intravenous, intramuscular,
subcutaneous, and intraperitoneal administration. Preferably, sterile aqueous media
are used, as is known to those skilled in the art, selected particularly in light
of the present description. Preferably, chemical buffers, which are buffering or regulating
solutions that maintain the pH of the composition stable, are used. Examples of buffers
are PBS. By way of illustration, a single dose can be dissolved in 1 ml of isotonic
NaCl solution and added to 1000 ml of hypodermoclysis fluid or injected into the proposed
infusion site (see, for example, "
Remington Pharmaceutical Sciences" 15th edition, pages 1035-1038 and 1570-1580). Some variations in dosage will necessarily occur depending on the condition of
the subject being treated. The person responsible for administration will, in any
case, determine the suitable dosage for the individual subject. Moreover, for human
administration, preparations must meet biological standards of sterility, pyrogenicity,
general safety, and purity as required, for example, by the ICH Quality Guidelines
cited above or by FDA regulations.
[0090] Furthermore, the significant increase of MBNL1 and/or MBNL2 proteins demonstrated
in the present invention leads to consider the possible use of these antagonists as
a drug. Therefore, a
third aspect of the invention relates to the antagonist according to the first aspect or any of
its embodiments, or to the composition according to the second aspect or any of its
embodiments, for use thereof as a drug. In a preferred embodiment, the use is as a
treatment for myotonic dystrophy, preferably myotonic dystrophy type 1.
[0091] The administration of the antagonist by means of a possible expression vector expressing
same can allow directing the expression to a specific tissue or group of tissues depending
on the tropism of the base vector itself and/or by means of the choice of control
elements that result in the expression of the coding sequence linked to same only
in specific tissues. As for the organs or tissues to be treated, preferably they are
those tissues or organs affected by DM1, in particular those selected from the group
of skeletal muscle, cardiac muscle, smooth muscles, diaphragm, respiratory muscles,
or pharyngeal muscles or stem cells of one or more of said organs.
[0092] Furthermore, particular dosage forms may favor greater access to one or more other
organs. The concentration assays performed in the examples show that the antagomiRs,
at different doses, are able to increase the expression and concentration of MBNL1
and/or MBNL2 proteins, which supports their use for the preparation of a drug for
the treatment of myotonic dystrophy 1, in particular to palliate symptoms of the disease,
especially symptoms corresponding to muscle dysfunction. In one possible embodiment,
the pharmaceutical composition comprises an effective dose of an inhibitor or antagonist
of at least one of the human microRNAs miR-100, miR-20, miR-222, miR-181, and miR-92.
More preferably, the inhibitor(s)/antagonist(s) will be present at a concentration
that allows the administration of a therapeutically effective dose.
[0093] An "effective dose" or "therapeutically effective dose" is an amount sufficient to
effect a beneficial or desired clinical outcome. Furthermore, the effective dose may
vary in organisms of different species and/or sizes. Thus, an effective dose of an
inhibitor/antagonist of a microRNA in rodents, in accordance with previous results
obtained with molecules targeting other microRNAs, can be from about 1 mg/kg to about
100 mg/kg, about 2.5 mg/kg to about 50 mg/kg, or about 5 mg/kg to about 25 mg/kg.
It should be noted that the therapeutic doses tested in mice must be converted to
their human equivalent. For this purpose, the person skilled in the art may consult
the well-known common practice guide for dose conversion between animals and human,
see, for example,
Nair AB, Jacob S. A simple practice guide for dose conversion between animals and
human. J Basic Clin Pharma 2016;7:27-31. Briefly, therapeutic doses in mouse models can be converted to humans by dividing
by about 12.3, although a corresponding clinical trial would have to be performed.
Calculating and obtaining an effective dose for humans is within routine practice
and therefore within the knowledge of one skilled in the art. Furthermore, the precise
determination of what is considered an effective dose may be based on individual factors
for each patient, including their size, age, and the nature of the inhibitor or antagonist
(e.g., whether it is an expression construct, an antagomiR or antimiR oligoribonucleotide
analog). Therefore, dosages can be readily determined by those skilled in the art
from this description and knowledge of the art. It may be necessary or desirable to
administer multiple doses to the subject during a particular treatment period, administering
doses daily, weekly, monthly, every two months, every three months, or every six months.
In certain embodiments, the subject receives an initial dose at a first time point
that is greater than one or more subsequent or maintenance doses.
[0094] Given the stability of antagomiRs, it is possible to consider their administration
to humans directly, for example by subcutaneous or systemic route, preferably by intravenous
route, for example dissolved or suspended in a pharmaceutically acceptable vehicle,
such as water or some aqueous solution such as, for example, saline or phosphate buffer.
[0095] Each embodiment described here is contemplated as being applicable to each of the
other described embodiments. Therefore, all the combinations of the various elements
described here are within the scope of the invention.
[0096] The following clauses also are included in the present invention:
- 1. An antagonist of human microRNA miR-100 capable of increasing the intracellular
levels of MBNL1 and/or MBNL2 proteins.
- 2. The antagonist of human microRNA miR-100 according to clause 1, wherein the intracellular
levels of MBNL1 and/or MBNL2 proteins are increased in a muscle cell, preferably in
a muscle cell having a muscular dystrophy phenotype, preferably myotonic dystrophy
type 1.
- 3. The antagonist of human microRNA miR-100 according to any of clauses 1 or 2, wherein
said antagonist is a molecule of an oligonucleotide nature and/or an oligonucleotide
analog.
- 4. The antagonist of human microRNA miR-100 according to any of clauses 1 to 3, wherein
said antagonist is an antimiR, a blockmiR, or a microRNA sponge.
- 5. The antagonist of human microRNA miR-100 according to any of clauses 1 to 4, wherein
said antagonist comprises a region which is at least 85% identical to the complementary
sequence of SEQ ID NO: 1.
- 6. The antagonist of human microRNA miR-100 according to any of clauses 1 to 5, wherein
said antagonist comprises a region of at least 7 contiguous nucleotide or nucleotide
analog units which is 100% identical to the sequence complementary to the seed region
of human microRNA miR-100 as defined in SEQ ID NO: 2.
- 7. The antagonist of human microRNA miR-100 according to any of clauses 1 to 6, wherein
said antagonist comprises a first region of at least 7 contiguous nucleotide or nucleotide
analog units which is 100% identical to the sequence complementary to the seed region
of human microRNA miR-100 as defined in SEQ ID NO: 2, and wherein said antagonist
further comprises a second region of at least 7 contiguous nucleotide or nucleotide
analog units adjacent to the first region, wherein said second region is at least
90% identical to the sequence complementary to any region present in SEQ ID NO: 1.
- 8. The antagonist of human microRNA miR-100 according to any of clauses 1 to 9, wherein
said antagonist comprises a nucleotide or nucleotide analog region which is at least
90% identical to SEQ ID NO: 3.
- 9. The antagonist of human microRNA miR-100 according to any of clauses 1 to 8, wherein:
- i) said antagonist comprises a region which is at least 95% identical to SEQ ID NO:
3,
- ii) at least one of the nucleotide or nucleotide analog units making up said antagonist
is a nucleotide analog having one or more chemical modifications in the ribose moiety,
in the phosphate bond, or in both, and
- iii) optionally, said antagonist has at the 5' end and/or at the 3' end one or more
additional molecules, preferably cholesterol.
- 10. The antagonist of human microRNA miR-100 according to any of clauses 1 to 9, wherein
said antagonist comprises a nucleotide or nucleotide analog region which is at least
95% identical to any of the sequences SEQ ID NO: 8 or 13.
- 11. The antagonist of human microRNA miR-100 according to any of clauses 9 or 10,
wherein said antagonist consists of SEQ ID NO: 8 or SEQ ID NO: 13.
- 12. A composition comprising at least one antagonist of human microRNA miR-100 as
defined in any of clauses 1 to 11.
- 13. The composition according to clause 12, wherein said composition is a pharmaceutical
composition further comprising a pharmaceutically acceptable vehicle and/or one or
more pharmaceutically acceptable excipients.
- 14. The antagonist according to any of clauses 1 to 11, or composition according to
any of clauses 12 or 13, for use thereof as a drug.
- 15. The antagonist according to any of clauses 1 to 11, or composition according to
any of clauses 12 or 13, for use thereof in the treatment of myotonic dystrophy, preferably
myotonic dystrophy type 1.
SEQUENCE LISTING
[0097] Sequence of human microRNA miR-100-5p: SEQ ID NO 1:
AACCCGUAGAUCCGAACUUGUG
[0098] Seed sequence of human microRNA miR-100-5p: SEQ ID NO 2: CCCGUAG
[0099] Sequence of antagonist hsa-miR-100: SEQ ID NO 3:
CACAAGTTCGGATCTACGGGTT
[0100] Sequence of antagonist hsa-miR-20a-5p: SEQ ID NO 4:
CTACCTGCACTATAAGCACTTTA
[0101] Sequence of antagonist hsa-miR-181c-5p: SEQ ID NO 5:
ACTCACCGACAGGTTGAATGTT
[0102] Sequence of antagonist hsa-miR-222-3p: SEQ ID NO 6:
ACCCAGTAGCCAGATGTAGCT
[0103] Sequence of antagonist hsa-miR-92a-3p: SEQ ID NO 7:
ACAGGCCGGGACAAGTGCAATA
[0104] Sequence of antagonist hsa-miR-100-5p with modifications: SEQ ID NO 8:
C*A*CAAGTTCGGATCTACGG*G*T*T*
[0105] Sequence of antagonist hsa-miR-20a-5p with modifications: SEQ ID NO 9:
C*T*ACCTGCACTATAAGCACT*T*T*A*
[0106] Sequence of antagonist hsa-miR-181c-5p with modifications: SEQ ID NO 10:
A*C*TCACCGACAGGTTGAAT*G*T*T*
[0107] Sequence of antagonist hsa-miR-222-3p with modifications: SEQ ID NO 11:
A*C*CCAGTAGCCAGATGTA*G*C*T*
[0108] Sequence of antagonist hsa-miR-92a-3p with modifications: SEQ ID NO 12:
A*C*AGGCCGGGACAAGTGCA*A*T*A* where:
- an asterisk (*) indicates the presence of a phosphorothioate bond,
- A, C, T, G in bold indicates that said nucleotide is modified with 2'-OMe, and
- optionally, a fatty acid is conjugated at the 3' and/or 5' ends of the molecule.
[0109] Sequence of antagonist hsa-miR-100-5p with modifications: SEQ ID NO 13:
C*A*CAAGTTCGGATCTACGG*G*T*T*-Chol
[0110] Sequence of antagonist hsa-miR-20a-5p with modifications: SEQ ID NO 22:
C*T*ACCTGCACTATAAGCACT*T*T*A*-Chol
[0111] Sequence of antagonist hsa-miR-181c-5p with modifications: SEQ ID NO 23:
A*C*TCACCGACAGGTTGAAT*G*T*T*-Chol
[0112] Sequence of antagonist hsa-miR-222-3p with modifications: SEQ ID NO 24:
A*C*CCAGTAGCCAGATGTA*G*C*T*-Chol
[0113] Sequence of antagonist hsa-miR-92a-3p with modifications: SEQ ID NO 25:
A*C*AGGCCGGGACAAGTGCA*A*T*A*-Chol
where:
- an asterisk (*) indicates the presence of a phosphorothioate bond,
- A, C, T, G in bold indicates that said nucleotide is modified with 2'-OMe, and
- -Chol indicates that the oligonucleotide is conjugated to cholesterol.
[0114] Sequence of human microRNA miR-20a-5p: SEQ ID NO 14:
UAAAGUGCUUAUAGUGCAGGUAG
[0115] Sequence of human microRNA miR-222-3p: SEQ ID NO 15:
AGCUACAUCUGGCUACUGGGU
[0116] Sequence of human microRNA miR-181c-5p: SEQ ID NO 16:
AACAUUCAACCUGUCGGUGAGU
[0117] Sequence of human microRNA miR-92a-3p: SEQ ID NO 17:
UAUUGCACUUGUCCCGGCCUGU
[0118] Seed sequence of human microRNA miR-20a-5p SEQ ID NO 18: AAGUGCU
[0119] Seed sequence of human microRNA miR-222-3p: SEQ ID NO 19: CUACAUC
[0120] Seed sequence of human microRNA miR-181c-5p: SEQ ID NO 20: CAUUCAA
[0121] Seed sequence of human microRNA miR-92a-3p: SEQ ID NO 21: UUGCACU
[0122] Sequence of antagonist hsa-miR-100-5p without modifications: SEQ ID NO 26:
CACAAGTTCGGATCTACGGGTT
[0123] Sequence of antagonist hsa-miR-20a-5p without modifications: SEQ ID NO 27:
CTACCTGCACTATAAGCACTTTA
[0124] Sequence of antagonist hsa-miR-181c-5p without modifications: SEQ ID NO 28:
ACTCACCGACAGGTTGAATGTT
[0125] Sequence of antagonist hsa-miR-222-3p without modifications: SEQ ID NO 29:
ACCCAGTAGCCAGATGTAGCT
[0126] Sequence of antagonist hsa-miR-92a-3p without modifications: SEQ ID NO 30:
ACAGGCCGGGACAAGTGCAATA
[0127] The invention will now be illustrated in further detail with the help of the examples
and figures which are shown below.
EXAMPLES
EXAMPLE 1: Screening based on mimetic microRNA libraries (SureFIND Transcriptome PCR
Array, Qiagen).
[0128] This is a method for the identification of regulators of gene expression.
[0129] In this study, the "Cancer miRNA SureFind Transcriptome PCR Array" kit was used to
identify potential miRNA regulators of MBNL1 and 2. These arrays include cDNA from
HeLa cells treated with defined miRNA mimetics in 96-well plate format (90 different
microRNAs and 6 controls) in which the target mRNAs of the miRNAs are expected to
be detected significantly reduced or increased by real-time PCR. Multiplex qPCR assay
was performed using commercial Taqman probes (QuantiFast Probe PCR Kits, Qiagen) to
quantify the expression of MBNL1 and 2 (genes of interest, labeled with FAM) and GADPH
(as reference endogenous expression, labeled with MAX). Changes in the expression
of MBNL1 and 2 as a result of treatment with each specific mimetic microRNA were calculated
with respect to the mimetic negative control (a microRNA that does not exist in nature)
and normalized with respect to GADPH. The observed changes were plotted as log2 and
subjected to
ΔΔCt (MAD) statistical analysis for the selection of positive candidate miRNAs (Figure
1).
Bioinformatic 3'UTR binding predictions
[0130] Starting from those microRNAs that had repressor activity on MBNL1 and MBNL2 at the
messenger level, the inventors searched for possible targets of these microRNAs that
bind directly to the 3'UTR of MBNL1 and MBNL2. For this purpose, 2 databases were
used, mirDIP and miRecords, which collect information from a total of 9 prediction
programs including TargetScan and miRanda, providing information on the existence
of targets of a specific microRNA in the transcripts of a given gene suggesting direct
regulation.
Cell culture toxicity assay (TC50)
[0131] DM1 fibroblasts were cultured in Dulbecco's Modified Eagle Medium-high glucose (DMEM
4500 mg/l, Gibco) supplemented with 1% penicillin/streptomycin and 10% inactivated
fetal bovine serum in cell culture bottles. Given their adherent growth, for their
passage, they were washed with PBS and trypsinized for 2 min at 37°C, then fresh medium
was added to inhibit the action of trypsin.
[0132] Cells for this assay were seeded at a density of 10
5 cells/ml in 96-well plates (10,000 cells per well). The plate was seeded according
to the following template. In the case of column 1 (blank), no cells were seeded,
as this column is the target of the colorimetry analysis. The rest of the plate was
seeded with DM1 fibroblasts, which were subsequently treated as described below. About
16 hours after cell seeding and with the cells at 80% confluence, the cells were transfected
with the commercial antagomiRs provided by Biomers (antagomiR-miR-100-5p (SEQ ID NO:
3): CACAAGTTCGGATCTACGGGTT, antagomiR-20a-5p (SEQ ID NO: 4): CTACCTGCACTATAAGCACTTTA,
antagomiR-181c-5p (SEQ ID NO: 5): ACTCACCGACGACAGGTTGAATGTT, antagomiR-222-3p (SEQ
ID NO: 6): ACCCAGTAGCCAGATGTAGCT and antagomiR-miR-92a-3p (SEQ ID NO: 7): ACAGGCCCCGGGGGACAAGTGCAATA)
using X-tremeGENE HP reagent (Roche) according to the manufacturer's instructions.

[0133] All the antagomiRs were transfected into patient fibroblasts (Arandel
et al. 2017) using increasing amounts of 10 nM, 50 nM, 200 nM, and 1000 nM, and 5000 nM
(C1, C2, C3, C4, and C5, respectively), and as a control only transfection reagent
but no antagomiR was introduced, the transfection medium was left together with the
cells for 4 hours, and after this time it was changed to transdifferentiation medium.
To transdifferentiate fibroblasts to myoblasts, MyoD expression was induced. To that
end, the complete medium was replaced with muscle differentiation medium (MDM) consisting
of DMEM supplemented with 1% P/S, 2% horse serum (Gibco), 0.1 mg/ml apotransferrin,
0.01 mg/ml insulin, and 0.02 mg/ml doxycycline (Sigma) for 60 h.
[0134] After 72 h, the transdifferentiation medium was replaced with 100 µl of fresh medium
in all wells of the plate including column 1, and 20 µl of the MTS/PMS solution (CellTiter
96
® Aqueous Non-Radioactive Cell Proliferation Assay Kit) were added to each well and
incubated for 2 h at 37°C. After the incubation time, the colorimetric assay was read
in the Infinite 200 PRO Microplate Reader, Tecan, following the manufacturer's instructions.
The data obtained with the reader was processed and analyzed in order to obtain the
TC50 which allows knowing the amount of antagomiR to be used for it not to be toxic
in the cell model (Figure 2).
Assay of the activity of antagomiR on MBNL1 protein (EC50)
[0135] Fibroblasts from DM1 patients and healthy controls were cultured in Dulbecco's Modified
Eagle Medium-high glucose (DMEM 4500 mg/l, Gibco) supplemented with 1% P/S and 10%
inactivated fetal bovine serum in cell culture bottles. The cells were seeded in 6-well
plates, at a density of 100,000 cells/well by introducing 2.5 ml of cells in each
well. About 16 hours after cell seeding and with the cells at 80% confluence, the
cells were transfected with the commercial antagomiRs provided by Biomers (antagomiR-miR-100-5p
(SEQ ID NO: 3): CACAAGTTCGGATCTACGGGTT, antagomiR-20a-5p (SEQ ID NO: 4): CTACCTGCACTATAAGCACTTTA,
antagomiR-181c-5p (SEQ ID NO: 11): CTCACCGACGACAGGTTGAATGTT, antagomiR-222-3p (SEQ
ID NO: 6): ACCCAGTAGCCAGATGTAGCT and antagomiR-miR-92a-3p (SEQ ID NO: 7): ACAGGCCCCGGGGGACAAGTGCAATA),
using X-tremeGENE HP reagent (Roche).
[0136] The antagomiRs were transfected into patient fibroblasts using increasing amounts
of 10 nM, 50 nM, 200 nM, 1000 nM, and 5000 nM. As a control only transfection reagent
but no antagomiRs were introduced into both healthy and DM1 cells. Subsequently the
transfection medium was left together with the cells for 4 hours and after this time
it was changed to transdifferentiation medium (DMEM supplemented with 1% P/S, 2% horse
serum (Gibco), 0.1 mg/ml apotransferrin, 0.01 mg/ml insulin, and 0.02 mg/ml doxycycline
(Sigma)). Fibroblasts were transdifferentiated to myoblasts for 72 hours.
[0137] After 72 h, total protein used for QDB assays was extracted using RIPA buffer (Thermo
Scientific) plus protease and phosphatase inhibitors (Roche Applied Science). Samples
were quantified by BCA protein assay kit (Pierce) using bovine serum albumin as a
standard. Samples were prepared at a concentration of 1 µg/well along with 4x loading
buffer (0.8 ml 3 M Tris-HCl pH 6.8, 4 ml glycerol, 0.8 g SDS, 0.04 g bromophenol blue,
4 ml β-mercaptoethanol, ddH
2O up to 10 ml). Samples were denatured by boiling at 100°C for 5 min and loaded onto
QDB plates (Quanticision Diagnostics Inc). Each sample was loaded in quadruplicate
on two different plates (5 µl/well), one to detect MBNL1 and the other to detect GAPDH,
used as endogenous control, following the protocol described in Moreno-Cervera
et al., 2022. After this, they were left to dry and were incubated with transfer buffer
(14.4 g glycine, 3 g Tris base, 700 ml ddH
2O, 200 ml methanol, ddH
2O up to 1 I) under stirring for 1 min. This was followed by three washes with 1x TBS-T
(100 ml 10x TBS, 5 ml Tween 20, ddH
2O up to 1 l) and incubated for 1 hour with blocking buffer (5 g skim milk powder,
1x TBS-T up to 100 ml). Plates were incubated at 4°C overnight with anti-MBNL1 (1:1000,
ab77017, Abcam) and anti-GAPDH (1:500, clone G-9, Santa Cruz) primary antibodies.
All primary antibodies were detected using HRP (horseradish peroxidase)-conjugated
anti-mouse IgG secondary antibody at a concentration of 1:5000 (Sigma-Aldrich). Immunoreactivity
was detected using Pierce ECL Western Blotting Substrate reagent (Thermo Scientific)
and chemiluminescence levels were determined with the Inifinite M200 Pro plate reader
(Tecan).
Expression of candidate miRNAs in the relevant tissues
[0138] The expression levels of miR-100-5p, miR-20a-5p, miR-181c-5p miR-222-3p, and miR-92a-3p
in transcripts per million (TPM), from the different tissues of interest (trachea,
thyroid, testicles, stomach, spleen, different types of muscle cells, skeletal muscle,
liver, kidney, heart, bladder, esophagus, diaphragm, colon, cervix, brain, and blood)
were obtained by means of using the database, which is created by means of data from
the project. Fantom hosts a large number of samples of more than 100 different types
of tissue, therefore those which may be of interest for the disease have been filtered.
Name |
TC50 |
EC50 |
Emax |
TIndex |
AntagomiR-23b |
0.806 |
0.0278 |
2.596 |
75.254 |
AntagomiR-218 |
0.848 |
0.0224 |
1.870 |
70.810 |
AntagomiR-20a |
0.340 |
0.0100 |
1.458 |
49.572 |
AntagomiR-92a |
1.201 |
0.4170 |
2.434 |
7.010 |
AntagomiR-100 |
1.524 |
0.0180 |
1.552 |
131.622 |
AntagomiR-181c |
3.849 |
0.4675 |
1.340 |
11.034 |
AntagomiR-222 |
0.579 |
0.9777 |
1.979 |
1.172 |
[0139] Therapeutic index
of the different oligonucleotides used in DM1 cells. TC50, EC50, Emax, and therapeutic index parameters obtained for each of the antagomiRs
tested in DM1 cells.
Expression of candidate miRNAs in disease model cells
[0140] The relative expression levels of microRNAs miR-23b, miR-218, miR-92a, mirR-100,
miR-181c, miR-222, miR-20a, miR-17, and miR-let7a were measured by means of RT-qPCR
both in control cells and in DM1 model cells after 4 days of differentiation. Levels
of miR-1 were also measured as a positive control, since it is described (Rau
et al., 2011) that miR-1 is reduced in DM1 cells. All the levels were obtained using U1,
U6, and miR-103b as normalizers.
|
Screening mkroRNAS |
No. 3'UTR binding prediction programs |
qPCR Validation |
Expression in tissues |
Overexpression in DM1 |
miRs |
↓MB NL1 |
↓MBNL2 |
MBNL1 |
MBNL2 |
TarBase v7.0 |
↓MBNL 1 |
↓MBNL 2 |
muscle |
myotubes |
hsa-miR-20a |
Yes |
Yes |
5 |
4 |
|
|
|
Yes |
Yes |
hsa-miR-181c |
|
|
8 |
|
+ |
Yes |
|
Yes |
Yes |
hsa-miR-222 |
Yes |
Yes |
4 |
4 |
+ |
|
|
Yes |
Yes |
hisa-miR-100 |
Yes |
Yes |
6 |
|
|
|
|
Yes |
Yes |
hsa-miR-92a |
Yes |
Yes |
3 |
3 |
+ |
|
|
Yes |
Yes |
[0141] Table 3. Compilation of data obtained on the different miRNAs under study. The different parameters of interest analyzed in the study are shown: confirmation
of MBNL1 and 2 reduction in the overexpression screening of the miRNAs under study;
number of programs predicting the binding of miRNAs to MBNL1 and 2 and confirmation
by TarBase v7.0; validation of MBNL1 and 2 reduction by qPCR when the miRNAs of interest
are overexpressed; confirmation of miRNA expression in tissues of interest for DM1
(muscle); and confirmation of miRNA overexpression in DM1 myotubes model of the disease.
[0142] As can be seen in Table 2, antagomiR-100 showed the best Tindex values with respect
to the rest of the antagomiRs, which shows that this antagomiR presents the best balance
between toxicity, Emax, and EC50. However, it should also be noted that antagomiR-20b
has good EC50 and Emax values, antagomiR-181c has a good Emax value, and antagomiR-92a
has good TC50 and Emax values comparable to antagomiR-23b. Note that low EC50 values
indicate higher antagonistic potency of the miRNA in question, while higher TC50 indicates
lower toxicity on the cell model of the disease.
[0143] Furthermore, protein expression levels were studied in the cell model of the disease
(Figure 3) finding, firstly, that DM1 muscle cells express significantly less MBNL1
protein than the control. Taking as a reference the expression detected in DM1 cells,
it was found that in all cases an increase in total protein levels occurs, although
some antagomiRs are active at lower concentrations than others and the levels of activation
may be different. For example, for antagomiR-20a or -100, at 10 nM (the lowest concentration
tested), recovery in MBNL1 expression reaches normal levels, whereas other antagomiRs
are able to double expression in DM1 cells (antagomiR-92a and -222). This indicates
that some antagomiRs are more effective than others, such as antagomiR-181c, the activation
of MBNL1 expression of which is discrete, and requires higher concentration to achieve
levels similar to those of the rest of the molecules.
[0144] Figure 4 shows the evaluation of the activity and toxicity of the different antagomiRs
in DM1 cells after transfection with same at the different concentrations indicated.
The different graphs visually show the Tindex metric given for those antagomiRs for
which the EC and TC curves are farther apart; these are the ones that that show the
greatest difference between the concentrations at which they are active and those
at which they show toxic effects (therapeutic window). For example, antagomiR-100
shows activity at low concentrations and rapidly reaches the maximum possible activity,
while only at concentrations almost two orders of magnitude higher do they show a
reduction by half in the viability of DM1 muscle cells. This behavior is similar to
that of antagomiR-23b and -218, which are shown in comparison. In contrast, antagomiR-222
can increase MBNL1 expression markedly, but does so at concentrations at which the
toxicity curve is already above 50% dead cells. The rest of the antagomiRs show an
intermediate behavior. AntagomiR-20a also shows an interesting behavior since at the
lowest concentration tested it already reaches the maximum activity in terms of MBNL1
activation, with its TC50 being at least one order of magnitude higher. In antagomiR-181c,
the difference between activity and toxicity is low. Taking both curves together,
it is observed that antagomiR-100 presents a similar pattern to the previously patented
antagomiRs, -23b and -218, since its activity levels are very high from the second
dose tested, but its toxicity level exceeds the TC50 only at high doses, i.e., it
presents a wider therapeutic window and therefore is more suitable for pharmacological
development.
[0145] Next, the expression of the microRNAs of interest was determined in different human
tissues (Figure 5). As can be seen, microRNA miR-100 is expressed almost exclusively
in skeletal muscle and its satellite cells, with only some expression in fibroblasts,
which is of particular interest because skeletal muscle is a tissue that is greatly
affected in DM1, so that the antagonistic effect of antimiR-100 is expected to be
specific, avoiding possible undesirable consequences due to repression of this miRNA
in tissues that are not altered in the disease. For example, since antimiRs accumulate
in the liver and kidney, the fact that the antimiR-100 target is not expressed in
these organs will prevent reaching a lack of function situation of this miRNA in these
organs, which is desirable from a therapeutic point of view. On the other hand, the
expression of miR-100 in satellite cells is also of great interest because these are
the muscle stem cells from which muscle regeneration can be promoted. In fact, it
has been demonstrated (Song KY,
et al. MBNL1 reverses the proliferation defect of skeletal muscle satellite cells in myotonic
dystrophy type 1 by inhibiting autophagy via the mTOR pathway.
Cell Death Dis. 2020 Jul 18;11(7):545. doi: 10.1038/s41419-020-02756-8. PMID: 32683410;
PMCID: PMC7368861) that an increase of MBNL1 in these cells reverses the proliferative defects that
these cells have in DM1, so acting at the satellite cell level through a satellite
cell-specific target is anticipated to be potentially therapeutic
in vivo. Finally, the detection of miR-100 in mature skeletal muscle confirms the relevance
of the observations made
in vitro with cell models of the disease, which may differ markedly with the
in vivo expression pattern in adult humans.
[0146] Finally, an assay was performed to quantify possible differences in expression of
the different target miRNAs in cell models of the disease with respect to their healthy
controls. The results in Figure 6 show that most of the selected miRNAs are detected
overexpressed in an
in vitro model of the disease, with this characteristic being desirable in a therapeutic target
because it reduces the risk of harmful effects due to lack of target function. That
is, the preferable therapeutic target is not only to reduce the expression of an miRNA
below its normal values, but to bring it to levels similar to those of healthy controls,
thus preserving its natural endogenous functions. As a control, note that miR-218,
previously described as overexpressed in DM1 (see
Cerro-Herreros E et al. Preclinical characterization of antagomiR-218 as a potential
treatment for myotonic dystrophy. Mol Ther Nucleic Acids. 2021 Jul 29;26:174-191.
doi: 10.1016/j.omtn.2021.07.017. PMID: 34513303; PMCID: PMC8413838), is detected as being increased in this cell model, as well as miR-23b, while miR-17
and others do not show any significant change in expression and serve as a control
that this is not a general effect on all miRNAs analyzed.